ArticlePDF AvailableLiterature Review

Abstract and Figures

The novel coronavirus pandemic has taken a toll on the global healthcare systems and economy. Safety precautions, along with vaccination, are the most effective preventive measures. The global vaccination program against COVID-19 has dramatically reduced the number of deaths and cases. However, the incidence of thrombotic events and thrombocytopenia post-COVID-19 vaccination known as vaccine-induced thrombotic thrombocytopenia has raised safety concerns. This has led to an element of vaccine hesitancy. The exact mechanism for vaccine-induced thrombotic thrombocytopenia is unknown. Although the incidence of thrombosis associated with COVID-19 vaccination is low, it still requires attention, especially in older people, smokers, and people with preexisting comorbidities. This study aims to review the pathophysiology, diagnosis, and management of vaccine-induced thrombotic thrombocytopenia, to provide a concise and comprehensive update.
Content may be subject to copyright.
Journal Pre-proof
Vaccine-induced thrombotic thrombocytopenia following coronavirus vaccine: A
narrative review
Syed Hassan Ahmed, Taha Gul Shaikh, Summaiyya Waseem, Nashwa Abdul Qadir,
Zohaib Yousaf, Irfan Ullah
PII: S2049-0801(21)00938-9
DOI: https://doi.org/10.1016/j.amsu.2021.102988
Reference: AMSU 102988
To appear in: Annals of Medicine and Surgery
Received Date: 5 September 2021
Revised Date: 27 October 2021
Accepted Date: 28 October 2021
Please cite this article as: Ahmed SH, Shaikh TG, Waseem S, Qadir NA, Yousaf Z, Ullah I, Vaccine-
induced thrombotic thrombocytopenia following coronavirus vaccine: A narrative review, Annals of
Medicine and Surgery (2021), doi: https://doi.org/10.1016/j.amsu.2021.102988.
This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition
of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of
record. This version will undergo additional copyediting, typesetting and review before it is published
in its final form, but we are providing this version to give early visibility of the article. Please note that,
during the production process, errors may be discovered which could affect the content, and all legal
disclaimers that apply to the journal pertain.
© 2021 Published by Elsevier Ltd on behalf of IJS Publishing Group Ltd.
Vaccine-Induced Thrombotic Thrombocytopenia Following Coronavirus Vaccine: A Narrative Review
Syed Hassan Ahmed1, Taha Gul Shaikh1, Summaiyya Waseem1, Nashwa Abdul Qadir1, Zohaib Yousaf2,
Irfan Ullah3
1Dow University of Health Sciences, Karachi, Pakistan
2Department of Internal Medicine, Hamad Medical Corporation, Doha, Qatar
3Kabir Medical College, Gandhara University, Peshawar, Pakistan
Corresponding Author
Zohaib Yousaf (MBBS, MSc, FACP)
Department of Internal Medicine,
Hamad Medical Corporation, Doha, Qatar
Email: zohaib.yousaf@gmail.com
Irfan Ullah, MBBS
Kabir Medical College Gandhara University, Peshawar, 25000 Pakistan
Email: irfanullahecp2@gmail.com
Abstract= 121
Word Count= 2244
Tables and figure= 1 Table and 2 Figures
References= 65
Journal Pre-proof
Vaccine-Induced Thrombotic Thrombocytopenia Following Coronavirus Vaccine: A
Narrative Review
Abstract
The novel coronavirus pandemic has taken a toll on the global healthcare systems and
economy. Safety precautions, along with vaccination, are the most effective preventive
measures. The global vaccination program against COVID-19 has dramatically reduced the
number of deaths and cases. However, the incidence of thrombotic events and
thrombocytopenia post-COVID-19 vaccination known as vaccine-induced thrombotic
thrombocytopenia has raised safety concerns. This has led to an element of vaccine
hesitancy. The exact mechanism for vaccine-induced thrombotic thrombocytopenia is
unknown. Although the incidence of thrombosis associated with COVID-19 vaccination is low,
it still requires attention, especially in older people, smokers, and people with preexisting
comorbidities. This study aims to review the pathophysiology, diagnosis, and management of
vaccine-induced thrombotic thrombocytopenia, to provide a concise and comprehensive
update.
Key words: COVID-19, COVID-19 Vaccine, Vaccine Induced Thrombotic Thrombocytopenia,
Vaccine Induced Immune Thrombotic Thrombocytopenia, Thrombotic thrombocytopenia,
VITT
Journal Pre-proof
Introduction
The Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2) cases were initially
reported in Wuhan, China, towards the end of 2019. Following its extensive spread, the
World Health Organization (WHO) declared COVID-19 a pandemic in March 2020. [1] To the
date, April 16, approximately 207 million confirmed cases have been reported, and 4.3
million deaths [2].
Coordinated global efforts led to the development of COVID-19 vaccines, followed by
emergency use authorization within nine months of the pandemic [3]. These vaccines are
now widely available for public administration [4]. The vaccines are safe and effective in
preventing severe infection, hospitalization, and death [5,6]. To date, 4.4 billion vaccine doses
have been administered [2]. The common adverse effects following COVID-19 vaccination are
injection site pain and transient, self-limited systemic symptoms like headache, fever,
myalgias, etc. [7].
Recently, a more severe adverse effect, thrombocytopenia with or without thrombosis, has
been reported following SARS-CoV-2 vaccination. Thrombocytopenia is a medical condition
characterized by platelets lower than 150,000/microliter and is associated with a risk of
bleeding and thrombosis [8]. Such reports have raised concerns over the safety profile and
hesitancy towards the available vaccines [9]. The term "Vaccine-Induced Thrombotic
Thrombocytopenia" describes post-vaccination thrombocytopenia cases. VITT is
characterized by thrombosis at unusual sites and thrombocytopenia following vaccination [9].
While VITT has been associated with both mRNA and viral vector vaccines, its prevalence is
higher in viral vectored vaccines [7]. Following the incidence of 30 thromboembolism cases in
March 2021, Oxford/AstraZeneca (AZD1222) was transiently suspended in numerous
European countries [10]. Later the pharmacovigilance risk assessment committee (PRAC) of
the European medical agency (EMA) reviewed all cases and declared thrombosis and
thrombocytopenia as rare adverse effects of AZD1222. However, based on risk-benefit
assessment, the vaccine was later declared safe for use [11]. Owing to a similar reason, in
April 2021, Johnson & Johnson’s Janssen (Ad26.CoV2.S) administration was also temporarily
suspended [12].
Herein, we review the association between SARS-CoV-2 vaccines and VITT. This review
evaluates the potential pathophysiology and clinical approach to diagnoses and management
of VITT.
Literature Review
The work has been reported in line with the PRISMA 2020 criteria [13]. Two authors
(SHA, SW) dependently conducted a thorough literature search over PubMed and
Clinicaltrials.gov from inception till August 16, 2021, without any language
restriction. To achieve comprehensive results, search string comprised of keywords,
"SARS-CoV-2 Vaccine", "Coronavirus Vaccine," "Corona Vaccine," "COVID-19
Vaccine", "thrombotic thrombocytopenic," "Vaccine-Induced Thrombotic
Thrombocytopenia," "VITT," "thrombocytopenia," "reduced platelet count," using
BOOLEAN operators. Synonyms, related terms, and spelling variants were also
engaged. All relevant case reports, case series, cohort studies, editorials, and
correspondences were reviewed. Any discrepancies were resolved via discussion with
a third reviewer (IU). The results of the literature search are shown in figure 1.
Following studies selection, two independent authors (TGS, NAQ) extracted all the
Journal Pre-proof
relevant data into a table comprising of author's name, patient's age, and sex, past
medical history, presenting complaint, laboratory findings, radiological findings,
treatment interventions, and outcome. Any discrepancies were resolved by discussion
with a third reviewer (IU). All significant findings are summarized in table 1.
Figure 1: PRISMA Flowchart
Demographics
The retrieved studies comprise data of 44 patients (32 females, 11 males, 1 not defined) with
a mean age of 44.9 ± 14.3 years. The following figure (Figure 2) depicts the geographical
distribution of the reported cases around the globe, with the majority of cases arising in
Europe. Based on these and future reporting, we can predict the potential spatial spread,
geographical locations that may be more susceptible than others and this may help us
Journal Pre-proof
establish links between different genetic and environmental factors, predisposing an
individual to such consequences of vaccines.
Figure 2: Geographical Distribution of the Reported Cases
Pathophysiology
The exact pathophysiology behind VITT is unclear. As shown in Table 1, most of the cases
presented with thrombocytopenia, elevated D-dimer, and positive titers of IgG antibodies
against platelet factor 4 (PF-4)[1422]. Based on these findings, this syndrome is closely
related to heparin-induced thrombocytopenia (HIT), a medical condition characterized by
thrombocytopenia, and the presence of antibodies against the Heparin-PF4 complex [23].
HIT, an autoimmune reaction to heparin, involves the generation of IgG antibodies against
the Heparin-PF4 complex. The Fc portion of these antibodies adheres to the complex, binds
to the FcYRIIa receptors [24], and initiates platelets activation via intracellular signaling
involving spleen tyrosine kinase [25]. This results in the release of microparticles and a
procoagulant state [26,27]. Furthermore, clearance of activated and antibody-bound
platelets by the reticuloendothelial system culminates in thrombocytopenia [28]. A
prerequisite in the diagnosis of HIT includes a known recent exposure to heparin. A condition
labeled "Autoimmune Heparin-Induced Thrombocytopenia (aHIT)” manifests with clinical and
laboratory findings without any prior use of heparin [29]. Based on this resemblance, a
comparison has been drawn between VITT and variants of aHIT [30], and hence, we may
assume that a similar mechanism follows post-vaccination. However, the mechanism behind
the generation of these antibodies is yet to be elucidated.
In HIT, the electrostatic interaction between positively charged PF4 and negatively charged
heparin culminates in the formation of the Heparin-PF4 complex [31]. This phenomenon has
also been observed with other negatively charged molecules like numerous polyphosphates
[32], Polyvinyl phosphonate [33], nucleic acids [34], etc. According to Visentin et al. [33],
Journal Pre-proof
numerous negatively charged molecules, spaced about 0.5 nm apart along the molecular
backbone and of sufficient length, can form complexes with PF4 while being detectable by
the antibodies. Hence, components of vaccines can be expected to play a crucial role in the
generation of PF4-polyanions complex and antibodies against them. Moreover,
environmental factors and genetic predisposition can exacerbate clinical presentation. For
example, specific genotypes encoding FcRIIA have been associated with an increased risk of
thrombosis in individuals with anti-PF4-polyanion antibodies [24].
Another postulated mechanism involves cross-reactivity of anti-SARS-CoV-2 spike protein
antibodies that generates following SARS-CoV-2 vaccination with PF4. This may be
attributable to molecular mimicry, a phenomenon whereby a certain degree of resemblance
exists between the pathogens and the host's antigens [35]. Kanduc et al. [36] report massive
homogeneity between the SARS-CoV-2 spike glycoprotein and human proteins, thus further
strengthening this hypothesis. This structural resemblance can also explain the findings of
thrombocytopenia [37] and anti-PF4 antibodies [38] in certain SARS-CoV-2 patients. However,
the currently available literature suggests no evidence of cross-reactivity [38,39].
Zhang et al. [40] investigated the findings of thrombosis and thrombocytopenia in SARS-CoV-
2 patients. They reported spike protein's ability to stimulate platelet activation and thrombus
formation via the Mitogen-activated protein kinase (MAPK) pathway. Based on the findings
[40], the generation of spike protein following vaccination can also play a pivotal role in
inducing thrombocytopenia and thrombosis via spike protein-ACE2 interaction-induced
platelets activation. However, it remains unanswered if similar interactions can be observed
post-vaccination with vector or mRNA vaccines. Moreover, some evidence [41] reveals
potential interactions between adenovirus particles and circulating platelets leading to
platelet activation and aggregation. The possibility of such interactions in the case of viral
vector-based vaccines cannot be ruled out and requires further investigation. Furthermore,
as shown in table 1, the findings of negative anti-PF4 antibodies in selective cases indicate
involvement of a non-HIT like mechanism hence strengthening the above suggested
hypothesis.
Future research should focus on potential interactions between spike proteins and platelets
and the phenomenon of cross-reactivity. Another intriguing aspect of the higher prevalence
of VITT among individuals vaccinated with viral vector-based vaccines needs to be
investigated in the search for potential links. Development of thrombosis in selective
individuals and incidence of rare site thrombosis like cerebral venous sinuses deserve equal
attention for the exact pathophysiology to be elucidated. Lastly, the development of anti-PF4
antibodies only in certain VITT patients can also provide important clues in determining the
pathogenesis.
Diagnosis
Following the escalation in reported thrombocytopenia and thrombosis cases post COVID-19
vaccination, the American Society of Hematology (ASH) reviewed all the reported cases and
laid specific ground rules to diagnose this novel presentation. As per the ASH [42], cases
meeting the following criteria can be identified as VITT:
a) Symptom onset 4 to 42 days post SARS-CoV-2 vaccination
b) Any venous or arterial thrombosis (often cerebral or abdominal)
c) Thrombocytopenia
Journal Pre-proof
d) Antibodies to platelet factor 4 (PF4) identified by enzyme-linked immunosorbent test
(ELISA)
e) Markedly elevated D-dimer (> 4 times upper limit of normal)
Individuals presenting with the complaints of severe headache, visual changes, abdominal
pain, nausea, vomiting, back pain, shortness of breath, leg pain or swelling, petechiae, easy
bruising, or bleeding, 4 to 42 days post-vaccination, must be evaluated critically for the
condition mentioned above. Laboratory investigations, including CBC with platelet count, PF4
ELISA, d-dimer, fibrinogen, and imaging techniques for thrombosis, can play a crucial role in
timely diagnosis and management. [42]
Management
Currently, numerous potential pharmacological therapies are being evaluated in the line of
management for VITT. The outcomes range from being propitious to contraindicated or
variable in different individuals. Briefed below are specific interventions being employed to
overcome VITT.
Intravenous Immunoglobulins (IVIG)
The currently available evidence acknowledges IVIG as a potential treatment depicting
remarkable success. Hence it is now incorporated into the treatment regimen. A potential
explanation for this involves the Fcγ receptor blockade by the antibodies. The recommended
dose in VITT is 1-2g/kg of the person's body weight. However, ideally, the administered IVIG
should be the ones collected before the pandemic. The plausible explanation being vaccine
response deterioration due to COVID-19 antibodies present in the donated IGs [43].
Anticoagulants
There has been growing evidence of their efficacy in patients with VITT [44]. In some
instances, preliminary trials to validate its effectiveness and progressive clinical worsening in
some instances [45] have raised suspicions over its use regarding heparin. Therefore, the
American society of hematology (ASH) suggests avoiding the use of heparin unless VITT has
been ruled out or another condition diagnosed [42]
The drug of choice is direct oral anticoagulants (dabigatran, apixaban, rivaroxaban, edoxaban,
and fondaparinux) or parenteral direct thrombin inhibitors (e.g., bivalirudin and argatroban).
The absolute contraindication following anti-coagulation therapy includes a high risk of
bleeding. Hence strict clinical monitoring is crucial after initiating oral anticoagulants.
Steroids
Most cases of VITT described steroids as a clinically effective treatment option. However,
further data is needed to move past the anecdotal evidence. The above data and prediction
are based on their successfully reported usage in our included cases and recently, by Schultz
et al. [46], where the combined IVIG and steroids were supported.
Platelet Infusion
This therapy is only indicated in significant bleeding. Goel et al. [47] reported a five times
increase in mortality of patients infused with platelets following thrombocytopenia. In our
included studies, eight reportedly administered platelet infusion. Following Goel et al.
[47,48], only two patients survived [49].
Platelet Exchange
Journal Pre-proof
Plasma exchange was used in three cases. Two out of the three patients survived [50][51].
Relevant details by Garnier et al. were unavailable [14]. Plasma exchange is used in refractory
VITT [52]. Clinically, there is insufficient data to evaluate whether plasma exchange can be
administered safely in VITT.
Plasma exchange is not a standard treatment option in HIT [42]. Extrapolating this to VITT, we
may assume similar effects on patients with VITT. However, more data is required to draw
any conclusion.
Aspirin and Rituximab
Aspirin or other anti-platelets are currently contra-indicated in VITT due to increased risk of
bleeding. Smith et al. [53] suggested a possible prophylactic role of antiplatelets in VITT. This
highlights the need for more work in this area.
Rituximab is not recommended currently due to its longer response time (6-8 weeks) [42].
Moreover, this drug’s mechanism of action can be explained via its downregulation of CD-20
B-cells. This can potentially lead to the inactivation of antibodies against COVID-19, hence
rendering the vaccine administration useless.
Treatment Regimen
The following regimen is per the American Society of Hematology (ASH) [42], International
Society on Thrombosis and Haemostasis (ISTH) [54], and National Institute for Health and
Care Excellence (NICE) in the United Kingdom:
1. Start IVIG.
2. The ISTH guidelines recommend administering steroids if a patient's platelet count is
less than 50 x 109/L.
3. Platelet infusion and plasma exchange should not be considered initially.
4. Based on their history and previous clinical profile, patients shall be started an
anticoagulant (non-heparin). Vitamin K antagonists should be avoided while the
platelet count is low. Moreover, direct thrombin inhibitors should be avoided in
pregnant women. DOACs and fondaparinux are suitable for noncritically ill patients.
5. For patients having less than 50 ×103/μL and severe risk of bleeding, IV direct
thrombin inhibitors can be used. This will lead to a shorter half-life and rapid action.
6. Fibrinogen levels should be strictly monitored and kept in range (>1.5 g/L.)
7. If platelet count remains less than 30 x 109/L despite intravenous immunoglobulin
and steroid treatment or fibrinogen level is less than 1 g/L, plasma exchange can be
considered after an opinion with hematologists.
Conclusion
VITT is a rare adverse effect of SARS-CoV-2 vaccination, and the benefits of COVID-19
vaccines continue to outweigh the rare side effects. However, while its incidence is low, there
is undoubted an overwhelming need to discern the precise pathophysiology behind this
syndrome to establish proper management protocols. Questions like why certain coronavirus
vaccines carry a higher risk than others, why specific individuals develop thrombosis while
others don’t, higher prevalence in a particular gender and age group, and the impact of
different interventions in such patients need to be investigated before a clear conclusion can
be drawn. Lastly, future studies must take into consideration both pre-and post-vaccination
investigations to discern the role of any underlying condition.
Journal Pre-proof
Acknowledgments
None
Ethics statement
Not applicable
Funding
None
Conflict of interests
The authors declare that there is no conflict of interests
Provenance and peer review
Not commissioned, externally peer-reviewed
References
[1] Mahase E. Covid-19: WHO declares pandemic because of "alarming levels" of
spread, severity, and inaction. BMJ. 2020;368:m1036. Published 2020 Mar 12.
doi:10.1136/bmj.m1036
[2] Myanmar: WHO Coronavirus Disease (COVID-19) Dashboard With Vaccination
Data | WHO Coronavirus (COVID-19) Dashboard With Vaccination Data, (n.d.).
https://covid19.who.int/region/searo/country/mm (accessed August 27,
2021).
[3] FDA Takes Key Action in Fight Against COVID-19 By Issuing Emergency Use
Authorization for First COVID-19 Vaccine | FDA, (n.d.).
https://www.fda.gov/news-events/press-announcements/fda-takes-key-
action-fight-against-covid-19-issuing-emergency-use-authorization-first-covid-
19 (accessed August 27, 2021).
[4] Castells MC, Phillips EJ. Maintaining Safety with SARS-CoV-2 Vaccines. N Engl J
Med. 2021;384(7):643-649. doi:10.1056/NEJMra2035343
[5] Vaccine efficacy, effectiveness and protection, (n.d.).
https://www.who.int/news-room/feature-stories/detail/vaccine-efficacy-
effectiveness-and-protection (accessed August 27, 2021).
[6] Ramanathan K, Antognini D, Combes A, et al. Planning and provision of ECMO
services for severe ARDS during the COVID-19 pandemic and other outbreaks
of emerging infectious diseases. Lancet Respir Med. 2020;8(5):518-526.
doi:10.1016/S2213-2600(20)30121-1
[7] Hernández AF, Calina D, Poulas K, Docea AO, Tsatsakis AM. Safety of COVID-19
vaccines administered in the EU: Should we be concerned? [published
correction appears in Toxicol Rep. 2021 Sep 2;:]. Toxicol Rep. 2021;8:871-879.
doi:10.1016/j.toxrep.2021.04.003
Journal Pre-proof
[8] Jinna S, Khandhar PB. Thrombocytopenia. In: StatPearls. Treasure Island (FL):
StatPearls Publishing; July 25, 2021.
[9] Elalamy I, Gerotziafas G, Alamowitch S, et al. SARS-CoV-2 Vaccine and
Thrombosis: An Expert Consensus on Vaccine-Induced Immune Thrombotic
Thrombocytopenia. Thromb Haemost. 2021;121(8):982-991. doi:10.1055/a-
1499-0119
[10] Wise J. Covid-19: European countries suspend use of Oxford-AstraZeneca
vaccine after reports of blood clots. BMJ. 2021;372:n699. Published 2021 Mar
11. doi:10.1136/bmj.n699
[11] AstraZeneca’s COVID-19 vaccine: EMA finds possible link to very rare cases of
unusual blood clots with low blood platelets | European Medicines Agency,
(n.d.). https://www.ema.europa.eu/en/news/astrazenecas-covid-19-vaccine-
ema-finds-possible-link-very-rare-cases-unusual-blood-clots-low-blood
(accessed August 27, 2021).
[12] Shay DK. Safety Monitoring of the Janssen (Johnson & Johnson) COVID-19
VaccineUnited States, MarchApril 2021. MMWR. Morbidity and Mortality
Weekly Report. 2021;70.https://doi.org/10.15585/MMWR.MM7018E2.
[13] M.J. Page, J.E. McKenzie, P.M. Bossuyt, I. Boutron, T.C. Hoffmann, C.D. Mulrow, et
al. The PRISMA 2020 statement: an updated guideline for reporting systematic
reviews. International Journal of Surgery (2021):88;105906.
[14] Garnier M, Curado A, Billoir P, Barbay V, Demeyere M, Dacher JN. Imaging of
Oxford/AstraZeneca® COVID-19 vaccine-induced immune thrombotic
thrombocytopenia. Diagn Interv Imaging. 2021;102(10):649-650.
doi:10.1016/j.diii.2021.04.005
[15] Cliff-Patel N, Moncrieff L, Ziauddin V. Renal Vein Thrombosis and Pulmonary
Embolism Secondary to Vaccine-induced Thrombotic Thrombocytopenia (VITT).
Eur J Case Rep Intern Med. 2021;8(7):002692. Published 2021 Jun 30.
doi:10.12890/2021_002692
[16] Mehta PR, Apap Mangion S, Benger M, et al. Cerebral venous sinus thrombosis
and thrombocytopenia after COVID-19 vaccination - A report of two UK cases.
Brain Behav Immun. 2021;95:514-517. doi:10.1016/j.bbi.2021.04.006
[17] Suresh P, Petchey W. ChAdOx1 nCOV-19 vaccine-induced immune thrombotic
thrombocytopenia and cerebral venous sinus thrombosis (CVST). BMJ Case
Rep. 2021;14(6):e243931. Published 2021 Jun 16. doi:10.1136/bcr-2021-
243931
[18] Tiede A, Sachs UJ, Czwalinna A, et al. Prothrombotic immune
thrombocytopenia after COVID-19 vaccination. Blood. 2021;138(4):350-353.
doi:10.1182/blood.2021011958
[19] Bjørnstad-Tuveng TH, Rudjord A, Anker P. Fatal cerebral haemorrhage after
COVID-19 vaccine. Fatal hjerneblødning etter covid-19-vaksine. Tidsskr Nor
Laegeforen. 2021;141:10.4045/tidsskr.21.0312. Published 2021 Apr 29.
doi:10.4045/tidsskr.21.0312
Journal Pre-proof
[20] Yahyavi-Firouz-Abadi N, Naik RP. Cerebral venous sinus thrombosis associated
with vaccine-induced thrombotic thrombocytopenia [published online ahead of
print, 2021 Aug 1]. Neuroradiol J. 2021;19714009211036687.
doi:10.1177/19714009211036687
[21] Wolf ME, Luz B, Niehaus L, Bhogal P, Bäzner H, Henkes H. Thrombocytopenia
and Intracranial Venous Sinus Thrombosis after “COVID-19 Vaccine
AstraZeneca” Exposure. Journal of Clinical Medicine. 2021; 10(8):1599.
https://doi.org/10.3390/jcm10081599
[22] Tølbøll Sørensen AL, Rolland M, Hartmann J, et al. A case of thrombocytopenia
and multiple thromboses after vaccination with ChAdOx1 nCoV-19 against
SARS-CoV-2. Blood Adv. 2021;5(12):2569-2574.
doi:10.1182/bloodadvances.2021004904
[23] Hogan M, Berger JS. Heparin-induced thrombocytopenia (HIT): Review of
incidence, diagnosis, and management. Vasc Med. 2020;25(2):160-173.
doi:10.1177/1358863X19898253
[24] Rollin J, Pouplard C, Gruel Y. Risk factors for heparin-induced
thrombocytopenia: Focus on Fcγ receptors. Thromb Haemost.
2016;116(5):799-805. doi:10.1160/TH16-02-0109
[25] Reilly MP, Sinha U, André P, et al. PRT-060318, a novel Syk inhibitor, prevents
heparin-induced thrombocytopenia and thrombosis in a transgenic mouse
model. Blood. 2011;117(7):2241-2246. doi:10.1182/blood-2010-03-274969
[26] Nevzorova TA, Mordakhanova ER, Daminova AG, et al. Platelet factor 4-
containing immune complexes induce platelet activation followed by calpain-
dependent platelet death. Cell Death Discov. 2019;5:106. Published 2019 Jun
24. doi:10.1038/s41420-019-0188-0
[27] Campello E, Radu CM, Duner E, Lombardi AM, Spiezia L, Bendo R, Ferrari S,
Simioni P, Fabris F. Activated platelet‐derived and leukocyte‐derived circulating
microparticles and the risk of thrombosis in heparin‐induced
thrombocytopenia: a role for PF4‐bearing microparticles?. Cytometry Part B:
Clinical Cytometry. 2018 Mar;94(2):334-
41.https://doi.org/10.1002/CYTO.B.21507.
[28] Chong BH. Heparin‐induced thrombocytopenia. Journal of thrombosis and
haemostasis. 2003 Jul;1(7):1471-8.https://doi.org/10.1046/J.1538-
7836.2003.00270.X.
[29] Greinacher A, Selleng K, Warkentin TE. Autoimmune heparin‐induced
thrombocytopenia. Journal of Thrombosis and Haemostasis. 2017
Nov;15(11):2099-114.https://doi.org/10.1111/JTH.13813.
[30] Arepally GM. Heparin-induced thrombocytopenia. Blood. 2017;129(21):2864-
2872. doi:10.1182/blood-2016-11-709873
[31] Suvarna S, Espinasse B, Qi R, et al. Determinants of PF4/heparin
immunogenicity. Blood. 2007;110(13):4253-4260. doi:10.1182/blood-2007-08-
105098
Journal Pre-proof
[32] Cines DB, Yarovoi SV, Zaitsev SV, et al. Polyphosphate/platelet factor 4
complexes can mediate heparin-independent platelet activation in heparin-
induced thrombocytopenia. Blood Adv. 2016;1(1):62-74. Published 2016 Nov
22. doi:10.1182/bloodadvances.2016000877
[33] Visentin GP, Moghaddam M, Beery SE, McFarland JG, Aster RH. Heparin is not
required for detection of antibodies associated with heparin-induced
thrombocytopenia/thrombosis. J Lab Clin Med. 2001;138(1):22-31.
doi:10.1067/mlc.2001.115525
[34] Jaax ME, Krauel K, Marschall T, et al. Complex formation with nucleic acids and
aptamers alters the antigenic properties of platelet factor 4. Blood.
2013;122(2):272-281. doi:10.1182/blood-2013-01-478966
[35] Rose NR, Mackay IR. Molecular mimicry: a critical look at exemplary instances
in human diseases. Cell Mol Life Sci. 2000;57(4):542-551.
doi:10.1007/PL00000716
[36] Kanduc D, Shoenfeld Y. Molecular mimicry between SARS-CoV-2 spike
glycoprotein and mammalian proteomes: implications for the vaccine.
Immunol Res. 2020;68(5):310-313. doi:10.1007/s12026-020-09152-6
[37] Zulfiqar AA, Lorenzo-Villalba N, Hassler P, Andrès E. Immune
Thrombocytopenic Purpura in a Patient with Covid-19. N Engl J Med.
2020;382(18):e43. doi:10.1056/NEJMc2010472
[38] Althaus K, Möller P, Uzun G, et al. Antibody-mediated procoagulant platelets in
SARS-CoV-2-vaccination associated immune thrombotic thrombocytopenia.
Haematologica. 2021;106(8):2170-2179. Published 2021 Aug 1.
doi:10.3324/haematol.2021.279000
[39] Greinacher A, Selleng K, Mayerle J, Palankar R, Wesche J, Reiche S, Aebischer A,
Warkentin TE, Muenchhoff M, Hellmuth JC, Keppler OT. Anti-SARS-CoV-2 spike
protein and anti-platelet factor 4 antibody responses induced by COVID-19
disease and ChAdOx1 nCov-19 vaccination.https://doi.org/10.21203/rs.3.rs-
404769/v1.
[40] Zhang S, Liu Y, Wang X, et al. SARS-CoV-2 binds platelet ACE2 to enhance
thrombosis in COVID-19. J Hematol Oncol. 2020;13(1):120. Published 2020 Sep
4. doi:10.1186/s13045-020-00954-7
[41] Jin YY, Yu XN, Qu ZY, et al. Adenovirus type 3 induces platelet activation in
vitro. Mol Med Rep. 2014;9(1):370-374. doi:10.3892/mmr.2013.1805
[42] Vaccine-induced Thrombotic Thrombocytopenia (VITT) and COVID-19 Vaccines:
What Cardiovascular Clinicians Need to Know - American College of Cardiology,
(n.d.). https://www.acc.org/latest-in-
cardiology/articles/2021/04/01/01/42/vaccine-induced-thrombotic-
thrombocytopenia-vitt-and-covid-19-vaccines (accessed August 27, 2021).
[43] Cimolai N. Untangling the Intricacies of Infection, Thrombosis, Vaccination, and
Antiphospholipid Antibodies for COVID-19 [published online ahead of print,
2021 Jun 22]. SN Compr Clin Med. 2021;1-16. doi:10.1007/s42399-021-00992-3
Journal Pre-proof
[44] von Hundelshausen P, Lorenz R, Siess W, Weber C. Vaccine-Induced Immune
Thrombotic Thrombocytopenia (VITT): Targeting Pathomechanisms with
Bruton Tyrosine Kinase Inhibitors [published online ahead of print, 2021 Apr
13]. Thromb Haemost. 2021;10.1055/a-1481-3039. doi:10.1055/a-1481-3039
[45] COVID-19: Vaccine-induced immune thrombotic thrombocytopenia (VITT) -
UpToDate, (n.d.). https://www.uptodate.com/contents/covid-19-vaccine-
induced-immune-thrombotic-thrombocytopenia-vitt (accessed August 27,
2021).
[46] Schultz NH, Sørvoll IH, Michelsen AE, et al. Thrombosis and Thrombocytopenia
after ChAdOx1 nCoV-19 Vaccination. N Engl J Med. 2021;384(22):2124-2130.
doi:10.1056/NEJMoa2104882
[47] Goel R, Ness PM, Takemoto CM, Krishnamurti L, King KE, Tobian AA. Platelet
transfusions in platelet consumptive disorders are associated with arterial
thrombosis and in-hospital mortality. Blood. 2015;125(9):1470-1476.
doi:10.1182/blood-2014-10-605493
[48] Tarawneh O, Tarawneh H. Immune thrombocytopenia in a 22-year-old post
Covid-19 vaccine. Am J Hematol. 2021;96(5):E133-E134. doi:10.1002/ajh.26106
[49] Bano F, Badugama B, Chandra D. Thrombosis and thrombocytopaenia after
ChAdOx1 nCoV-19 vaccination: a single UK centre experience. BMJ Case Rep.
2021;14(7):e243894. Published 2021 Jul 13. doi:10.1136/bcr-2021-243894
[50] Xie C, Vincent L, Chadwick A, Peschl H. COVID-19 vaccine induced
prothrombotic immune thrombocytopenia. Eur Heart J. 2021;42(33):3206.
doi:10.1093/eurheartj/ehab237
[51] Ruhe J, Schnetzke U, Kentouche K, et al. Acquired thrombotic
thrombocytopenic purpura after first vaccination dose of BNT162b2 mRNA
COVID-19 vaccine [published online ahead of print, 2021 Jul 26]. Ann Hematol.
2021;1-3. doi:10.1007/s00277-021-04584-y
[52] Kes P, Bašić-Jukić N, Brunetta Gavranić B. Therapeutic plasma exchange in
clinical practice: long-term single centre experience. Prilozi. 2011;32(2):67-85.
[53] Smith MR. Rituximab (monoclonal anti-CD20 antibody): mechanisms of action
and resistance. Oncogene. 2003;22(47):7359-7368.
doi:10.1038/sj.onc.1206939
[54] The ISTH Releases Interim Guidance on Vaccine-Induced Immune Thrombotic
Thrombocytopenia (VITT) - International Society on Thrombosis and
Haemostasis, Inc., (n.d.). https://www.isth.org/news/561406/ (accessed
August 27, 2021).
[55] Al-Maqbali JS, Al Rasbi S, Kashoub MS, et al. A 59-Year-Old Woman with
Extensive Deep Vein Thrombosis and Pulmonary Thromboembolism 7 Days
Following a First Dose of the Pfizer-BioNTech BNT162b2 mRNA COVID-19
Vaccine. Am J Case Rep. 2021;22:e932946. Published 2021 Jun 12.
doi:10.12659/AJCR.932946
Journal Pre-proof
[56] Muir KL, Kallam A, Koepsell SA, Gundabolu K. Thrombotic Thrombocytopenia
after Ad26.COV2.S Vaccination. N Engl J Med. 2021;384(20):1964-1965.
doi:10.1056/NEJMc2105869
[57] JCPSP | Journal of College of Physicians and Surgeons Pakistan, (n.d.).
https://jcpsp.pk/article-detail/thrombotic-thrombocytopenia-and-central-
venous-sinus-thrombosis-post-covid19-vaccination-and-its-treatment-with-
heparin-alternatives (accessed October 21, 2021).
[58] Ramdeny S, Lang A, Al-Izzi S, Hung A, Anwar I, Kumar P. Management of a
patient with a rare congenital limb malformation syndrome after SARS-CoV-2
vaccine-induced thrombosis and thrombocytopenia (VITT) [published online
ahead of print, 2021 Jun 7]. Br J Haematol. 2021;10.1111/bjh.17619.
doi:10.1111/bjh.17619
[59] Wiedmann M, Skattør T, Stray-Pedersen A, et al. Vaccine Induced Immune
Thrombotic Thrombocytopenia Causing a Severe Form of Cerebral Venous
Thrombosis With High Fatality Rate: A Case Series. Front Neurol.
2021;12:721146. Published 2021 Jul 30. doi:10.3389/fneur.2021.721146
[60] Gessler F, Schmitz AK, Dubinski D, et al. Neurosurgical Considerations
Regarding Decompressive Craniectomy for Intracerebral Hemorrhage after
SARS-CoV-2-Vaccination in Vaccine Induced Thrombotic Thrombocytopenia-
VITT. J Clin Med. 2021;10(13):2777. Published 2021 Jun 24.
doi:10.3390/jcm10132777
[61] Dias L, Soares-Dos-Reis R, Meira J, et al. Cerebral Venous Thrombosis after
BNT162b2 mRNA SARS-CoV-2 vaccine. J Stroke Cerebrovasc Dis.
2021;30(8):105906. doi:10.1016/j.jstrokecerebrovasdis.2021.105906
[62] Guetl K, Gary T, Raggam RB, Schmid J, Wölfler A, Brodmann M. SARS-CoV-2
vaccine-induced immune thrombotic thrombocytopenia treated with
immunoglobulin and argatroban. Lancet. 2021;397(10293):e19.
doi:10.1016/S0140-6736(21)01238-1
[63] Malik B, Kalantary A, Rikabi K, Kunadi A. Pulmonary embolism, transient
ischaemic attack and thrombocytopenia after the Johnson & Johnson COVID-19
vaccine. BMJ Case Rep. 2021;14(7):e243975. Published 2021 Jul 14.
doi:10.1136/bcr-2021-243975
[64] D'Agostino V, Caranci F, Negro A, et al. A Rare Case of Cerebral Venous
Thrombosis and Disseminated Intravascular Coagulation Temporally Associated
to the COVID-19 Vaccine Administration. J Pers Med. 2021;11(4):285. Published
2021 Apr 8. doi:10.3390/jpm11040285
[65] Mauriello A, Scimeca M, Amelio I, et al. Thromboembolism after COVID-19
vaccine in patients with preexisting thrombocytopenia. Cell Death Dis. 2021;12(8):762.
Published 2021 Aug 3. doi:10.1038/s41419-021-04058-z
Journal Pre-proof
Journal Pre-proof
Highlights
The global vaccination program against COVID-19 has greatly reduced the number of deaths and
cases.
This review evaluates all the currently available literature and highlights potential
pathophysiology, clinical approach to diagnose VITT and its management.
The escalating incidence of thrombotic events and thrombocytopenia post-COVID-19
vaccination has raised concerns regarding the safety profile of available vaccines
VITT has been acknowledged as a rare adverse effect of SARS-CoV-2 vaccination, and the
benefits of COVID-19 vaccines continue to markedly outweigh the rare ramifications.
Journal Pre-proof
Annals of Medicine and Surgery
The following information is required for submission. Please note that failure to respond to these
questions/statements will mean your submission will be returned. If you have nothing to declare in any of
these categories then this should be stated.
Please state any conflicts of interest
All authors must disclose any financial and personal relationships with other people or organisations that
could inappropriately influence (bias) their work. Examples of potential conflicts of interest include
employment, consultancies, stock ownership, honoraria, paid expert testimony, patent
applications/registrations, and grants or other funding.
None
Please state any sources of funding for your research
All sources of funding should be declared as an acknowledgement at the end of the text. Authors should
declare the role of study sponsors, if any, in the collection, analysis and interpretation of data; in the
writing of the manuscript; and in the decision to submit the manuscript for publication. If the study
sponsors had no such involvement, the authors should so state.
None
Ethical Approval
Research studies involving patients require ethical approval. Please state whether approval has been
given, name the relevant ethics committee and the state the reference number for their judgement.
NA
Journal Pre-proof
Consent
Studies on patients or volunteers require ethics committee approval and fully informed written consent
which should be documented in the paper.
Authors must obtain written and signed consent to publish a case report from the patient (or, where
applicable, the patient's guardian or next of kin) prior to submission. We ask Authors to confirm as part of
the submission process that such consent has been obtained, and the manuscript must include a
statement to this effect in a consent section at the end of the manuscript, as follows: "Written informed
consent was obtained from the patient for publication of this case report and accompanying images. A
copy of the written consent is available for review by the Editor-in-Chief of this journal on request”.
Patients have a right to privacy. Patients’ and volunteers' names, initials, or hospital numbers should not
be used. Images of patients or volunteers should not be used unless the information is essential for
scientific purposes and explicit permission has been given as part of the consent. If such consent is
made subject to any conditions, the Editor in Chief must be made aware of all such conditions.
Even where consent has been given, identifying details should be omitted if they are not essential. If
identifying characteristics are altered to protect anonymity, such as in genetic pedigrees, authors should
provide assurance that alterations do not distort scientific meaning and editors should so note.
NA
Author contribution
Please specify the contribution of each author to the paper, e.g. study concept or design, data
collection, data analysis or interpretation, writing the paper, others, who have contributed in
other ways should be listed as contributors.
SHA, IU: Study concept or design
SHA, SW, TGS, NAQ and IU: Data collection, data analysis or interpretation, writing the paper
ZY: Critical revision of the article
Journal Pre-proof
Registration of Research Studies
In accordance with the Declaration of Helsinki 2013, all research involving human participants
has to be registered in a publicly accessible database. Please enter the name of the registry and
the unique identifying number (UIN) of your study.
You can register any type of research at http://www.researchregistry.com to obtain your UIN if
you have not already registered. This is mandatory for human studies only. Trials and certain
observational research can also be registered elsewhere such as: ClinicalTrials.gov or ISRCTN
or numerous other registries.
1. Name of the registry: NA
2. Unique Identifying number or registration ID: NA
3. Hyperlink to your specific registration (must be publicly accessible and will
be checked): NA
Guarantor
The Guarantor is the one or more people who accept full responsibility for the work and/or the
conduct of the study, had access to the data, and controlled the decision to publish
Zohaib Yousaf (MBBS, MSc, FACP)
Department of Internal Medicine,
Hamad Medical Corporation, Doha, Qatar
Email: zohaib.yousaf@gmail.com
Irfan Ullah, MBBS
Kabir Medical College, Gandhara University,
Peshawar, Pakistan
Irfanullahecp2@gmail.com
+923340968239
Journal Pre-proof
Journal Pre-proof
... CVT related to coronavirus disease 2019 (COVID-19) vaccines has been reported. Vaccine-induced immune thrombotic thrombocytopenia (VITT) is becoming recognized as an adverse event of the adenoviral vector vaccine (Sharifian-Dorche et al. 2021;Ahmed et al. 2022). In contrast, CVT after messenger RNA (mRNA) vaccination is rare (Cheng 2021;Dias et al. 2021;Fan et al. 2021;Yamaguchi et al. 2021;Zakaria et al 2021). ...
... Currently, two types of COVID-19 vaccines exist, adenoviral vector vaccines and mRNA vaccines. CVT, specifically VITT, in patients who have received adenoviral vector COVID-19 vaccines has been reported (Sharifian-Dorche et al. 2021;Ahmed et al. 2022). VITT is reported to resemble heparin-induced thrombocytopenia and has a high mortality rate of 39% (Sharifian-Dorche et al. 2021). ...
... VITT is reported to resemble heparin-induced thrombocytopenia and has a high mortality rate of 39% (Sharifian-Dorche et al. 2021). The time from vaccination to the onset of CVT was 4-42 days (Ahmed et al. 2022). In contrast, CVT after an mRNA vaccine is rare. ...
Article
Full-text available
Antithrombin deficiency is a high-risk factor for venous thromboembolism during pregnancy, whereas cerebral venous thrombosis is rare. Cerebral venous thrombosis related to coronavirus disease 2019 (COVID-19) vaccines has been reported; however, there are a few reports of cerebral venous thrombosis after a messenger RNA (mRNA) vaccination. A 25-year-old female in her sixth week of pregnancy presented with headache 24 days after BNT162b2 mRNA COVID-19 vaccination. The following day, she presented with altered sensorium and was diagnosed with severe cerebral venous thrombosis. She demonstrated heparin resistance and was found to have an inherited antithrombin deficiency. A heterozygous missense variant in SERPINC1 (c.379T>C, p.Cys127Arg, 'AT Morioka') was detected by DNA analysis. Despite intensive care with unfractionated heparin, antithrombin concentrate, and repeated endovascular treatments, she died on the sixth day of hospitalization. Cerebral venous thrombosis in pregnant women with an antithrombin deficiency can follow a rapid and fatal course. Treatment with unfractionated heparin and antithrombin concentrate may be ineffective in severe cerebral venous thrombosis cases with antithrombin deficiency. Early recognition of antithrombin deficiency and an immediate switch to other anticoagulants may be required. Although the association between cerebral venous thrombosis and the vaccine is uncertain, COVID-19 vaccinations may require careful evaluation for patients with prothrombic factors.
... En febrero del 2021 se anunció la aparición de unos casos de trombosis del seno venoso cerebral con trombocitopenia y anticuerpos contra el factor 4 plaquetario (anti-PF4) en individuos inmunizados con la vacuna de AstraZeneca, y se acuñó el término "trombocitopenia trombótica inmune inducida por vacunas" o VITT a esta entidad [35][36][37][38], la cual se define de acuerdo con la Asociación Americana de Hematología (ASH), si cumple los siguientes cinco criterios: 1) inicio de los síntomas entre 4 y 42 días posteriores a la vacunación; 2) presencia de cualquier trombosis venosa o arterial; 3) trombocitopenia con recuento de plaquetas <150.000 células/μL; 4) nivel del dímero D >4 veces el límite superior normal; y, 5) anticuerpos anti-PFA positivos por técni-ca de ELISA [39,40]. Si todos los cinco criterios están presentes, se confirma el diagnóstico, si solo hay cuatro, es probable [35,41]. ...
... Posteriormente, este síndrome también fue reportado en individuos inmunizados con la vacuna de Johnson & Johnson [35], pero continúa en su mayoría siendo producido por la vacuna de AstraZeneca, probablemente debido a su uso más masivo [36]. Esto tuvo como consecuencia que ambas vacunas fueran suspendidas por un tiempo por las entidades reguladoras a nivel mundial [40,42,43]. Por otra parte, con las vacunas basadas en mRNA (Pfizer y Moderna), los casos reportados son pocos hasta ahora [43], y algunos autores consideran que se relacionan a condiciones preexistentes como trombocitopenia inmune y hemofilia A [36]. ...
... Otro de los efectos secundarios observado tanto en pacientes con COVID-19 como en los vacunados, es la trombosis de la vena porta. A raíz de las coagulopatías asociadas a la vacunación, en particular con las vacunas a base de vectores adenovirales, se encuentran en la literatura varias series de casos y revisiones sistemáticas con y sin metaanálisis, todas con resultados similares [34,40,41,44,48,[53][54][55][56][57]. Por mencionar los resultados de algunas, en una revisión sistemática se evaluó el número de pacientes reportados con VITT y trombosis del seno venoso cerebral inducidas por las vacunas, en PubMed hasta mayo de 2021, encontrando 41 pacientes afectados luego de recibir la vacuna de AstraZeneca y 13 pacientes con la de Johnson y Johnson. ...
Article
Full-text available
El virus SARS-CoV-2 continúa infectando a millones de individuos en el mundo. Aunque los síntomas más frecuentes observados en los pacientes con COVID-19 son fiebre, fatiga y tos, en los casos severos la hipercoagulabilidad y la inflamación son dos condiciones que pueden producir complicaciones y causar daño en órganos, poniendo en riesgo la vida del paciente. Con el fin de clasificar a los pacientes durante el triaje, se han explorado diferentes marcadores hematológicos, incluidos el recuento de plaquetas, linfocitos y eosinófilos, y la relación neutrófilos/linfocitos, entre otros. Por su parte, para la evaluación de las coagulopatías, se vienen determinando marcadores como el dímero D y el fibrinógeno. En esta revisión se abordan las coagulopatías y los parámetros hematológicos en pacientes con COVID-19, al igual que las anormalidades en la coagulación como la trombocitopenia trombótica inmune inducida por las vacunas contra el SARS-CoV-2.
... Finally, heparin was received by 61 % of survivors and 50 % with thrombosis. Recent systematic reviews and meta-analyses have focused on the pathophysiology, diagnosis, and treatment [24]; CVST [25]; analysis posthoc [26]; a scoring system to predict mortality [8]; and adenoviralvector-based vaccines [27]. Therefore, clinical presentation, risk factors, therapeutic approaches, and outcomes in survivors and non-survivors are unclear. ...
... The first VITT case series was published in February 2021, and the ChAdOx1 nCoV-19 vaccine was the most frequently associated [30]. All data in this meta-analysis and previously published evidence currently reproduce similar results [8,[24][25][26]. In our study, individuals vaccinated with mRNA vaccines (BNT162b2 and mRNA-1273) presented with an earlier onset of AEFI (p 0.001), a shorter time window between the most recent dose and hospital admission (p 0.0001), and more severe thrombocytopenia than those immunized with viral vector vaccines. ...
Article
Full-text available
Vaccine-inducing immune thrombocytopenia, thrombosis, and bleeding emerge as infrequent and potential complications with mortality risk in healthy subjects. However, differences between survivors and non-survivors with SARS-CoV-2 vaccine-induced thrombotic thrombocytopenia (VITT) are unclear. Methods According to the PRISMA statement, we conducted a systematic review and meta-analysis, and the protocol was registered in PROSPERO. The main objective is to identify differences among survivors and non-survivors of SARS-CoV-2 VITT patients. We systematically searched through PubMed, Scopus, and Web of Science. We included cohorts, case series, and case reports. We classified bleeding complications according to the ISTH definition. Statistics: unpaired Student’s t-test or one-way ANOVA, Wilcoxon, and Kruskal-Wallis. Results We systematically searched from January 2021 to June 2021 and identified 51 studies that included 191 patients. Non-survivors had the most severe thrombocytopenia (p 0.02) and lower fibrinogen measurements (p 0.01). Subjects vaccinated with mRNA vaccines (BNT162b2 and mRNA-1273) had an earlier onset of adverse events following immunization (p 0.001). We identified a higher trend of overall thrombotic events (p 0.001) in recipients of viral mechanism-dependent vaccines (Table 2). Non-survivors with cerebral venous sinus thrombosis (CVST) had more severe thrombocytopenia (p 0.01) than survivors with CVST. Finally, 61 % of survivors and 50 % with thrombosis received heparin. Conclusion We identified more severe thrombocytopenia, lower fibrinogen measurements, and a higher trend of overall thrombotic events, including CVST and thrombotic storm, particularly with viral mechanisms-dependent vaccines in non-survivors VITT patients.
... The rollout of the ChAdOx-vectored SARS-CoV-2 vaccine provided evidence that this platform can be administered on a large scale with high effectiveness. The use of the Adenovirusvectored COVID-19 vaccine was limited following the detection of rare, serious adverse events such as Vaccineinduced Immune Thrombotic Thrombocytopenia [75]. Therefore, the occurrence of such events, though rare, should be monitored closely in any clinical studies of this vector. ...
Article
Full-text available
Lassa virus, the cause of deadly Lassa fever, is endemic in West Africa, where thousands of cases occur on an annual basis. Nigeria continues to report increasingly severe outbreaks of Lassa Fever each year and there are currently no approved vaccines or therapeutics for the prevention or treatment of Lassa Fever. Given the high burden of disease coupled with the potential for further escalation due to climate change the WHO has listed Lassa virus as a priority pathogen with the potential to cause widespread outbreaks. Several candidate vaccines have received support and have entered clinical trials with promising early results. This review focuses on the current state of vaccine and therapeutic development for LASV disease and the potential of these interventions to advance through clinical trials. The growing burden of LASV disease in Africa highlights the importance of advancing preclinical and clinical testing of vaccines and therapeutics to respond to the growing threat of LASV disease.
... This compares a report that found a recognised risk factor for thrombosis was present in 65% of cases. 21 with COVID-19 would be more likely to have a DVT compared to those without. Due to the small sample size of the COVID-19 population in our results we are unable to make any conclusions. ...
Article
Introduction After the COVID‐19 vaccination roll out began in March 2021 patients began presenting to a Victorian Emergency Department with lower limb pain following their vaccination. As a result, radiology requests for ultrasound examinations, to exclude post vaccination deep vein thrombosis (DVT) began appearing. Method A retrospective search of lower limb venous ultrasound examinations was undertaken for a 1‐year period from March 2021 to February 2022. Patients were included in the study if they had a clinical indication of having been referred following COVID‐19 vaccination. Bivariate analysis was conducted, using logistic regression, to determine the strength of association between independent variables (i.e., age, gender, vaccination status, and COVID‐19 diagnosis) and dependent variables (i.e., a diagnosis of DVT and superficial vein thrombosis [SVT]). Results The study found 1627 examinations had a lower limb venous ultrasound examination over the study period. Ultrasound was positive for acute DVT in 218/1627 examinations (13.4%). 104/1627 (6.4%) presented for ultrasound following COVID‐19 vaccination. Six of these were found to have acute lower limb DVT detected. The association between post vaccination and DVT was an odds ratio (OR) of 0.379 (95% CI 0.164–0.874, p = .023). All examinations that were diagnosed with acute thrombus had recently received AstraZeneca (AZ) (Vaxzevria, ChAdOx1‐S/nCoV‐19) vaccine. Four examinations were diagnosed with superficial vein thrombosis (SVT) post recent COVID‐19 vaccination. One patient was diagnosed with Vaccine Induced Thrombosis with Thrombocytopenia after their first dose of AZ. Conclusion This study found 6/1627 (0.37%) lower limb venous ultrasound examinations, over a 1‐year period, were positive for acute DVT after COVID‐19 vaccination. The results showed post vaccination examinations were less likely to be diagnosed with a DVT than the population referred who had not had recent vaccination.
... Another complication is thrombosis which has been documented in association with COVID-19 [215][216][217][218][219][220][221][222][223][224][225][226][227][228][229] and COVID-19 vaccination [230][231][232][233]. In case of COVID-19 vaccination, vaccine-induced immune thrombotic thrombocytopenia (VITT), which can lead to cerebral venous sinus thrombosis, has been found to be particularly associated with the adenovirus vector-based COVID-19 vaccines [234][235][236][237][238][239][240][241][242]. Another example are retinal artery/vein occlusions induced by SARS-CoV-2 infection [243][244][245][246][247][248][249][250][251][252][253][254][255][256][257][258][259][260][261][262] and COVID-19 vaccination , which can thus be considered part of the symptoms of COVID-19 and PCVS. ...
Article
Full-text available
Worldwide there have been over 760 million confirmed coronavirus disease 2019 (COVID-19) cases, and over 13 billion COVID-19 vaccine doses have been administered as of April 2023, according to the World Health Organization. An infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can lead to an acute disease, i.e. COVID-19, but also to a post-acute COVID-19 syndrome (PACS, "long COVID"). Currently, the side effects of COVID-19 vaccines are increasingly being noted and studied. Here, we summarise the currently available indications and discuss our conclusions that (i) these side effects have specific similarities and differences to acute COVID-19 and PACS, that (ii) a new term should be used to refer to these side effects (post-COVID-19 vaccination syndrome, PCVS, colloquially "post-COVIDvac-syndrome"), and that (iii) there is a need to distinguish between acute COVID-19 vaccination syndrome (ACVS) and post-acute COVID-19 vaccination syndrome (PACVS) - in analogy to acute COVID-19 and PACS ("long COVID"). Moreover, we address mixed forms of disease caused by natural SARS-CoV-2 infection and COVID-19 vaccination. We explain why it is important for medical diagnosis, care and research to use the new terms (PCVS, ACVS and PACVS) in order to avoid confusion and misinterpretation of the underlying causes of disease and to enable optimal medical therapy. We do not recommend to use the term "Post-Vac-Syndrome" as it is imprecise. The article also serves to address the current problem of "medical gaslighting" in relation to PACS and PCVS by raising awareness among the medical professionals and supplying appropriate terminology for disease.
... VITT is caused by immunoglobulin G molecules that recognize platelet factor 4 (PF4) bound to platelets, which eventually causes platelet activation and the stimulation of the coagulation system; these antibodies are detectable through a PF4 enzyme-linked immunosorbent assay (ELISA) [8,[18][19][20][21]. Although it is not caused by heparin exposure, the disease process is similar to heparin-induced thrombocytopenia (HIT) [5,[22][23][24][25][26]. The clinical profile of VITT has not been completely elucidated either, but Vaccines 2022, 10,1950 2 of 10 cases have involved various organ systems, such as cerebral veins, pulmonary arteries, portal veins, and peripheral veins. ...
Article
Full-text available
COVID-19 has significantly affected public health, social life, and economies worldwide. The only effective way to combat the pandemic is through vaccines. Although the vaccines have been in use for some time, safety concerns have still been raised. The most typical adverse effects of receiving a COVID-19 vaccine are localized reactions near the injection site, followed by general physical symptoms such as headaches, fatigue, muscle pain, and fever. Additionally, some people may experience VITT (vaccine-induced immune thrombotic thrombocytopenia), a rare side effect after vaccination. We present the case of a 60-year-old female patient that developed VITT-like symptoms with spleno-portal thrombosis and intestinal ischemia two weeks after the administration of the Ad26.COV2-S vaccine. Surgical treatment consisted of extensive bowel resection with end jejunostomy and feeding ileostomy. Two weeks after the first operation, a duodenal-ileal anastomosis was performed. The patient was discharged five weeks after the onset of the symptoms. Although some rare adverse effects are associated with the SARS-CoV-2 vaccines, the risk of hospitalization from these harmful effects is lower than the risk of hospitalization from COVID-19. Therefore, recognizing VITT is significant for ensuring the early treatment of clots and proper follow-up.
... However, some severe side-effects have been reported as well including splanchnic venous thrombosis [10] and vaccine-induced immune thrombotic thrombocytopenia [11]. There is a need to address them since reports of adverse events have been regarded as one of the leading motives behind vaccine hesitancy in low-and middle-income countries (LMICs) [12]. ...
Article
Full-text available
Introduction: The coronavirus pandemic has caused massive damage to global health care and the economy. The vaccination program has been paced around the globe to return as soon as possible to pre-COVID time. Although all the vaccines have been approved after the rigorous clinical and safety trials, some adverse effects have surfaced and are being reported from different parts of the world. One such side effect is chilblain-like lesions following the COVID vaccination. Chilblain lesions, also known as pernio, are an inflammatory condition usually affecting the acral regions of the body. It is mostly reported from cold and damp areas and has multiple causes associated with it. Objective: This study aims to review the publicly available data and to provide concise and comprehensive information as well as evaluate the potential pathology, clinical approach, and management of CLL post-vaccination. Methods: An extensive literature search over PubMed, Cochrane library, Google Scholar, and Clinicaltrails. gov from inception till 5th October 2021, without any restriction of language was carried out. All the recruited articles were reviewed, and their bibliographies were also screened for any relevant information. Results: 12 studies (10 case reports and 2 case series) were retrieved reporting the incidence of CLL post-vaccination. 8 studies reported incidence in female patients while 5 reported in males, with one study mentioning no gender. Moreover, most of them were either from Europe or the United States of America, except for two cases, reported from Turkey. Conclusions: Although the overall incidence of Chilblains following COVID-19 vaccination is low, there is still a strong need to find out the exact mechanism behind this to redefine the safety and administration criteria of the vaccines and to formulate a proper management protocol.
... The current pandemic of COVID-19 has caused a drastic effect on the healthcare system and global economy, and the vaccination of COVID-19, although reducing the death rate, may cause thrombosis (which is also a complication of many other infections) [9,113]. Thrombosis has gained attention as a deadly complication of respiratory viral infections also (like influenza and coronavirus) [8]. ...
Article
Full-text available
The purpose of this study was to find the biological propensities of the vegetable plant Pleurospermum candollei by investigating its phytochemical profile and biological activities. Phytochemical analysis was done by spectroscopic methods to investigate the amount of total polyphenols, and biological evaluation was done by the different antioxidant, enzyme inhibitory (tyrosinase, α-amylase, and α-glucosidase), thrombolytic, and antibacterial activities. The highest amount of total phenolic and flavonoid contents was observed in methanolic extract (240.69 ± 2.94 mg GAE/g and 167.59 ± 3.47 mg QE/g); the fractions showed comparatively less quantity (57.02 ± 1.31 to 144.02 ± 2.11 mg GAE/g, and 48.21 ± 0.75 to 96.58 ± 2.30 mg QE/g). The effect of these bioactive contents was also related to biological activities. GCMS analysis led to the identification of bioactive compounds with different biological effects from methanolic extract (antioxidant; 55.07%, antimicrobial; 56.41%), while the identified compounds from the n-hexane fraction with antioxidant properties constituted 67.86%, and those with antimicrobial effects constituted 82.95%; however, the synergetic effect of polyphenols may also have contributed to the highest value of biological activities of methanolic extract. Molecular docking was also performed to understand the relationship of identified secondary metabolites with enzyme-inhibitory activities. The thrombolytic activity was also significant (40.18 ± 1.80 to 57.15 ± 1.10 % clot lysis) in comparison with streptokinase (78.5 ± 1.53 to 82.34 ± 1.25% clot lysis). Methanolic extract also showed good activity against Gram-positive strains of bacteria, and the highest activity was observed against Bacillus subtilis. The findings of this study will improve our knowledge of phytochemistry, and biological activities of P. candollei, which seems to be a ray of hope to design formulations of natural products for the improvement of health and prevention of chronic diseases; however, further research may address the development of novel drugs for use in pharmaceuticals.
Article
Full-text available
In 2019, the discovery of a new strain of Coronavirus, later referred to as SARS-CoV2 took the world by storm, leading to a pandemic and shutting down all global activities. Several measures were taken adequately to combat the viral havoc, including developing numerous vaccines. All the vaccines currently available for the general population went through rigorous screenings and trials to ensure maximum safety and were only approved after that. However, once they were rolled out in the markets and administered to the population, some adverse reactions were reported, one of which included uveitis. It is an ocular inflammatory condition of the uveal tract, choroid, or iris. If untreated, it can lead to severe consequences, including blindness. It is further divided into four categories based on its anatomical location. Despite the rare incidence of uveitis following COVID-19 vaccination, it may contribute to vaccine hesitancy; hence addressing and digging into the pathophysiological cause is crucial. This study evaluates all the pathophysiological and demographical links between COVID-19 vaccination and uveitis, suggesting appropriate management plans.
Article
Full-text available
While vaccination is the single most effective intervention to drastically reduce severe disease and death following SARS-CoV-2 infection, as shown in UK and Israel, some serious concerns have been raised for an unusual adverse drug reaction (ADR), including vaccine-induced immune thrombotic thrombocytopenia (VITT) with concurrent low platelets as well as capillary leak syndrome. In fact, the overall safety of the vaccine is highlighted by the low frequency of ADR considering that in UK, by the early June, 40 million first doses and 29 million second doses have been injected; nonetheless, 390 thrombotic events, including 71 fatal events have been reported. Interestingly, the cases reported low platelet counts with the presence of anti-platelet factor-4 (PF4) antibodies, indicating an abnormal clotting reaction. Here, out of three referred cases, we report a post-vaccine clinical case of fatal thrombosis with postmortem examination and whole exome sequencing (WES) analysis, whose pathogenesis appeared associated to a preexisting condition of thrombocytopenia due to myelodysplasia.
Article
Full-text available
During a 2-week period, we have encountered five cases presenting with the combination of cerebral venous thrombosis (CVT), intracerebral hemorrhage and thrombocytopenia. A clinical hallmark was the rapid and severe progression of disease in spite of maximum treatment efforts, resulting in fatal outcome in for 4 out of 5 patients. All cases had received ChAdOx1 nCov-19 vaccine 1–2 weeks earlier and developed a characteristic syndrome thereafter. The rapid progressive clinical course and high fatality rate of CVT in combination with thrombocytopenia in such a cluster and in otherwise healthy adults is a recent phenomenon. Cerebral autopsy findings were those of venous hemorrhagic infarctions and thrombi in dural venous sinuses, including thrombus material apparently rich in thrombocytes, leukocytes and fibrin. Vessel walls were free of inflammation. Extra-cerebral manifestations included leech-like thrombi in large veins, fibrin clots in small venules and scattered hemorrhages on skin and membranes. CVT with thrombocytopenia after adenovirus vectored COVID-19 vaccination is a new clinical syndrome that needs to be recognized by clinicians, is challenging to treat and seems associated with a high mortality rate.
Article
Full-text available
As with past illnesses, an approach has been taken to vaccinate the population and halt the spread of COVID-19. On 13 April 2021, the US Food and Drug Administration called for a halt in the administration of the Johnson & Johnson (J&J) COVID-19 vaccine due to reports of thrombosis and thrombocytopenia being associated with vaccination. We present the case of a 43-year-old woman with a history of dyslipidaemia, depression, gastro-oesophageal reflux disease and obesity presenting with dyspnoea, headache and light headedness of 3 days’ duration. Ten days prior, she had received the J&J COVID-19 vaccine. She was found to have thrombocytopenia, elevated D-dimers, pulmonary emboli and presented 1 day after discharge with an arterial clot despite being on apixaban. Six other US-based cases of venous thrombotic events are being reviewed at present. Patients should be informed of the possibility of such events to provide informed consent.
Article
Full-text available
The Medicines and Healthcare products Regulatory Agency (MHRA) of the UK has approved the use of three vaccines to combat COVID-19 (SARS-CoV-2). There have been rare reports of thrombosis after vaccination with the AstraZeneca vaccine. We present three cases of vaccine-induced thrombotic thrombocytopenia (VITT) in one UK district general hospital following administration of this vaccine. Two of the patients had asymptomatic pulmonary emboli, while the other is the first known case of both renal vein thrombosis and pulmonary embolism. Learning points: Vaccine-induced thrombotic thrombocytopenia (VITT) can be associated with unusual and multiple sites of thrombosis.Clinicians should have a low threshold for requesting anti-PF4 antibody tests and imaging (especially pulmonary imaging) in thrombocytopenic patients after administration of the AstraZeneca vaccine.We describe a localised cluster of VITT despite its rarity according to current statistics, highlighting the need for an efficient data collection system to ensure the incidence of VITT is accurately reported.
Article
Full-text available
Given the ongoing global SARS-CoV-2-vaccination efforts, clinical awareness needs to be raised regarding the possibility of an increased incidence of SARS-CoV-2-vaccine-related immune-mediated thrombocytopenia in patients with intracerebral hemorrhage (ICH) secondary to cerebral sinus and vein thrombosis (CVT) requiring (emergency) neurosurgical treatment in the context of vaccine-induced immune thrombotic thrombocytopenia (VITT). Only recently, an association of vaccinations and cerebral sinus and vein thrombosis has been described. In a number of cases, neurosurgical treatment is warranted for these patients and special considerations are warranted when addressing the perioperative coagulation. We, herein, describe the past management of patients with VITT and established a literature-guided algorithm for the treatment of patients when addressing the impaired coagulation in these patients. Increasing insights addressing the pathophysiology of SARS-CoV-2-vaccine-related immune-mediated thrombocytopenia guide physicians in developing an interdisciplinary algorithm taking into account the special considerations of this disease.
Article
Full-text available
Advanced SARS-CoV-2 infections not uncommonly associate with the occurrence of silent or manifest thrombotic events which may be found as focal or systemic disease. Given the potential complexity of COVID-19 illnesses, a multifactorial causation is likely, but several studies have focused on infection-induced coagulopathy. Procoagulant states are commonly found in association with the finding of antiphospholipid antibodies. The correlation of the latter with thrombosis and/or clinical severity remains controversial. Although measures of antiphospholipid antibodies most commonly include assessments for lupus anticoagulant, anticardiolipin, and anti-ß2-glycoprotein-I antibodies, lesser common antibodies have been detected, and there remains speculation that other yet undiscovered autoimmune thrombotic events may yet be found. The recent discovery of post-vaccination thromboses associated with platelet factor 4 antibody has created another level of concern. The pathogenesis of antiphospholipid antibodies and their role in COVID-19-related thrombosis deserves further attention. The multifactorial nature of thrombosis associated with both infection and vaccination should continue to be studied as new events unfold. Even if a cause-and-effect relationship is variable at best, such dedicated research is likely to generate other valuable insights that are applicable to medicine generally.
Article
Thrombosis-thrombocytopenia syndrome and cerebral venous sinus thrombosis have been rarely reported in patients who have received severe acute respiratory syndrome coronavirus 2 adenoviral vector vaccines. Awareness of this potential adverse effect, recognizing early clinical symptoms and subtle signs of cerebral venous sinus thrombosis on head computed tomography and brain magnetic resonance imaging, appropriate vascular imaging, and unique treatment for this condition is critical. This is a report of a case of vaccine-induced thrombotic thrombocytopenia and associated cerebral venous sinus thrombosis with emphasis on imaging and clinical course.
Article
We report clinical findings of three patients presenting with thrombosis and thrombocytopaenia 10–16 days following the first dose of the ChAdOx1 nCoV-19 vaccine against SARS-CoV-2. All patients presented to a major university teaching hospital in the UK over a 5-day period and were found to have high-titre antibodies against platelet factor 4 (PF4) without previous exposure to heparin. All three patients presented with extensive venous thrombosis, significant thrombocytopaenia, elevated D-dimer and borderline low fibrinogen. Two had fatal intracerebral haemorrhage secondary to cavernous venous sinus thrombosis and one had PE. Reference laboratory testing of serum demonstrated anti-PF4 antibodies in all three patients. The clinical and laboratory findings confirmed vaccine-induced thrombotic thrombocytopaenia (VITT) which was poorly described at the time of presentation. We were able to manage successfully one patient with PE with intravenous immunoglobulin and corticosteroids.
Article
A series of cases with rare thromboembolic incidents including cerebral sinus vein thrombosis (some of them fatal) and concomitant thrombocytopenia occurring shortly after vaccination with the COVID-19 vaccine AZD1222 (Vaxzevria) has caused significant concern and led to its temporary suspension in many countries. Immediate laboratory efforts in four of these patients have identified a tentative pathomechanism underlying this syndrome termed vaccine-induced prothrombotic immune thrombocytopenia (VIPIT) or vaccine-induced thrombosis with thrombocytopenia (VITT), which encompasses the presence of platelet-activating antibodies to platelet-factor 4/heparin complexes, possibly emulated by polyanionic constituents of AZD1222, and thus resembles heparin-induced thrombocytopenia (HIT). Because these immune complexes bind and activate platelets via Fcγ-receptor IIA (FcγRIIA), high-dose intravenous immunoglobulin G has been suggested for treatment of VIPIT in addition to non-heparin anticoagulants. Here we propose inhibitors of Bruton tyrosine kinase (Btk) approved for B-cell malignancies (e.g. ibrutinib) as another therapeutic option in VIPIT, as they are expected to pleiotropically target multiple pathways downstream of FcγRIIA-mediated Btk activation, e.g. as demonstrated for the effective inhibition of platelet aggregation, dense granule secretion, P-selectin expression and platelet-neutrophil aggregate formation stimulated by FcγRIIA cross-linking. Moreover, CLEC-2- and GPIb-mediated platelet activation, the interactions and activation of monocytes and the release of neutrophil extracellular traps, as encountered in HIT, could be attenuated by Btk inhibitors. As a paradigm for emergency repurposing of approved drugs in COVID-19, off-label use of Btk inhibitors in a low dose range not affecting haemostatic functions could thus be considered as a sufficiently safe option to treat VIPIT.